scispace - formally typeset
Search or ask a question
Posted ContentDOI

Plitidepsin has a positive therapeutic index in adult patients with COVID-19 requiring hospitalization

TL;DR: In this article, the effect of Plitidepsin on the antiviral effect of drugs was investigated using a clinical trial conducted by Pharmamar, S.A. and IrsiCaixa (Madrid, Spain).
Abstract: This study has been funded by Pharmamar, S.A. (Madrid, Spain). This work was supported by grants from the Government of Spain (PIE_INTRAMURAL_ LINEA 1 - 202020E079; PIE_INTRAMURAL_CSIC-202020E043). The research of CBIG consortium (constituted by IRTA-CReSA, BSC, & IrsiCaixa) is supported by Grifols pharmaceutical. We also acknowledge the crowdfunding initiative #Yomecorono (https://www.yomecorono.com). N.I.U. has non-restrictive funding from PharmaMar to study the antiviral effect of Plitidepsin. N.J.K. was funded by grants from the National Institutes of Health (P50AI150476, U19AI135990, U19AI135972, R01AI143292, R01AI120694, and P01AI063302); by the Excellence in Research Award (ERA) from the Laboratory for Genomics Research (LGR), a collaboration between UCSF, UCB, and GSK (#133122P); by the Roddenberry Foundation, and gifts from QCRG philanthropic donors. This work was supported by the Defense Advanced Research Projects Agency (DARPA) under Cooperative Agreement #HR0011-19-2-0020. The views, opinions, and/or findings contained in this material are those of the authors and should not be interpreted as representing the official views or policies of the Department of Defense or the U.S. Government. This research was partly funded by CRIP (Center for Research for Influenza Pathogenesis), a NIAID supported Center of Excellence for Influenza Research and Surveillance (CEIRS, contract # HHSN272201400008C), by DARPA grant HR0011-19-2-0020, by supplements to NIAID grants U19AI142733, U19AI135972 and DoD grant W81XWH-20-1-0270, and by the generous support of the JPB Foundation, the Open Philanthropy Project (research grant 2020-215611 (5384)), and anonymous donors to AG-S. S.Y. received funding from a Swiss National Foundation (SNF) Early Postdoc Mobility fellowship (P2GEP3_184202).

Summary (1 min read)

Jump to: [Introduction][Results][Discussion] and [References and Notes]

Introduction

  • As of April 2021, there have been approximately 135 million confirmed cases of COVID-19 reported to the World Health Organization (WHO), including 3 million deaths.
  • (6) Thus, the sea may prove to be a reservoir of potent, naturally selected antiviral compounds.
  • (7, 8) Plitidepsin targets eukaryotic translation elongation factor 1A (eEF1A) (9), which is one of the most abundant protein synthesis factors in the eukaryotic cell, (9, 10) and a protein known to be used by viruses to facilitate their replication inside their host.
  • (12, 13) Originally developed as a cancer treatment, plitidepsin has undergone an extensive clinical development program.
  • Specifically, several phase I and II clinical trials have been conducted to 20 explore different intravenous dosing schedules and infusion times (14-18).

Results

  • The authors have evaluated the antiviral activity of plitidepsin against different coronavirus species, 30 strains and variants (Materials and Methods).
  • (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
  • Forty-four patients completed the study through day 31, with one patient in the 1.5 mg cohort withdrawing from the study after the first infusion due to a grade 3 hypersensitivity reaction.
  • Baseline viral load was found to be significantly correlated with hospital discharge by Day 15, by logistic and Cox regression models.

Discussion

  • Since late 2019, the world has been coping with a global health threat caused by the novel 35 coronavirus, SARS-CoV-2.
  • In the set of preclinical studies of this and previous reports, plitidepsin showed strong antiviral activity and a positive therapeutic index in in vitro models of SARS-CoV-2 infection, with better performance than other drugs, including remdesivir (10, 28).
  • The copyright holder for this preprintthis version posted May 25, 2021.

References and Notes

  • World Health Organization W. WHO COVID-19 Dashboard.
  • Phase I 45 and pharmacokinetic study of aplidine, a new marine cyclodepsipeptide in patients with advanced malignancies.
  • (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
  • SARS-CoV-2 15 viral load is associated with increased disease severity and mortality.
  • Bertsimas D, Lukin G, Mingardi L, Nohadani O, Orfanoudaki A, Stellato B, et al. COVID-19 mortality risk assessment:.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
Plitidepsin has a positive therapeutic index in adult patients with COVID-19
requiring hospitalization
Authors: José F. Varona
1,2*
, Pedro Landete
3,4
, Jose A. Lopez-Martin
5
, Vicente Estrada
6,7
, Roger
Paredes
8,9
, Pablo Guisado-Vasco
10,11
, Lucía Fernández de Orueta
11,12
, Miguel Torralba
13,14
, Jesús
5
Fortún
15
, Roberto Vates
12
, José Barberán
1,2
, Bonaventura Clotet
8,9,16,17
, Julio Ancochea
3,4,18
,
Daniel Carnevali
10,11
, Noemí Cabello
19
, Lourdes Porras
20
, Paloma Gijón
21
, Alfonso Monereo
12
,
Daniel Abad
11,12
, Sonia Zúñiga
22
, Isabel Sola
22
, Jordi Rodon
23
, Nuria Izquierdo-Useros
24,25
,
Salvador Fudio
26
, María José Pontes
27
, Beatriz de Rivas
27
, Patricia Girón de Velasco
5
, Belén
Sopesén
5,28,29
, Antonio Nieto
30
, Javier Gómez
30
, Pablo Avilés
31
, Rubin Lubomirov
26
, Kris M.
10
White
32,33
, Romel Rosales
32,33
, Soner Yildiz
32,33
, Ann-Kathrin Reuschl
34
, Lucy G. Thorne
34
,
Clare Jolly
34
, Greg J. Towers
34
, Lorena Zuliani-Alvarez
35,36,37,38
, Mehdi Bouhaddou
35,36,37,38
,
Kirsten Obernier
35,36,37,38
, Luis Enjuanes
22
, Jose M. Fernández-Sousa
39
, Plitidepsin COVID - 19
Study Group
, Nevan J. Krogan
32,35,36,37,38*
, José M. Jimeno
5†
, Adolfo García-Sastre
32,33,40,41†*
.
Affiliations:
15
1
Departamento de Medicina Interna, Hospital Universitario HM Monteprincipe, HM
Hospitales, Madrid, Spain.
2
Facultad de Medicina, Universidad San Pablo-CEU, Madrid, Spain.
3
Hospital Universitario La Princesa, Madrid, Spain.
4
Universidad Autónoma de Madrid, Madrid, Spain.
20
5
Virology & Inflammation Unit, PharmaMar, S.A., Colmenar Viejo, Madrid, Spain.
6
Hospital Clínico San Carlos, Madrid, Spain.
7
Universidad Complutense de Madrid, Madrid, Spain.
8
Infectious Diseases Department, irsiCaixa AIDS Research Institute, Badalona, Barcelona,
Spain.
25
9
Hospital Germans Trias I Pujol, Badalona, Barcelona, Spain.
10
Internal Medicine Department. Hospital Universitario Quironsalud Madrid, Madrid, Spain.
11
Universidad Europea, Madrid, Spain.
12
Internal Medicine Department, Hospital Universitario de Getafe, Madrid, Spain.
13
Health Sciences Faculty, University of Alcalá, Madrid, Spain.
30
14
Guadalajara University Hospital, Guadalajara, Spain.
15
Hospital Universitario Ramón y Cajal, Madrid, Spain.
16
Universitat Autònoma de Barcelona, Barcelona, Spain.
17
Universitat de Vic, Universitat Central de Catalunya Barcelona, Spain.
18
Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de
35
Salud Carlos III (ISCIII), Madrid
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted May 25, 2021. ; https://doi.org/10.1101/2021.05.25.21257505doi: medRxiv preprint
NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.

2
19
Infectious Diseases Department, Clinico San Carlos University Hospital, Madrid, Spain.
20
Internal Medicine, Hospital General de Ciudad Real, Ciudad Real, Spain.
21
Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario
Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
22
Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-
5
CSIC), Madrid, Spain.
23
IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB,
Bellaterra, Spain
24
IrsiCaixa AIDS Research Institute, Badalona, Barcelona, Spain.
25
Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelone, Spain.
10
25
Clinical Pharmacology Unit, PharmaMar, Colmenar Viejo, Madrid, Spain.
27
Medical Affairs, PharmaMar, Colmenar Viejo, Madrid, Spain.
28
Sylentis, S.A.U., Tres Cantos, Madrid, Spain.
29
Biocross, S.L., Valladolid, Spain
30
Bio Statistics Unit, PharmaMar, Colmenar Viejo, Madrid, Spain.
15
31
Preclinical Unit, Pharmamar, Colmenar Viejo, Madrid, Spain.
32
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY,
USA.
33
Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai,
New York, NY, USA.
20
34
Division of Infection and Immunity, University College London, London WC1E 6BT,
United Kingdom.
35
Quantitative Biosciences Institute (QBI), University of California San Francisco, San
Francisco, CA 94158, USA.
36
J. David Gladstone Institutes, San Francisco, CA 94158, USA.
25
37
Quantitative Biosciences Institute (QBI), Coronavirus Research Group (QCRG), San
Francisco, CA 94158, USA.
38
Department of Cellular and Molecular Pharmacology, University of California, San
Francisco, CA 94518, USA.
39
Pharmamar, Colmenar Viejo, Madrid, Spain.
30
40
Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at
Mount Sinai, New York, NY, USA.
41
Tish Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
* Corresponding author. Email: jfvarona@hmhospitales.com (J.F.V.); Adolfo.Garcia-
Sastre@mssm.edu (A.G-S.); Nevan.Krogan@ucsf.edu, (N.J.K.)
35
These authors contributed equally to this work.
‡ A full list of the authors and affiliations included in the Supplementary Materials.
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted May 25, 2021. ; https://doi.org/10.1101/2021.05.25.21257505doi: medRxiv preprint

3
Abstract: Plitidepsin is a marine-derived cyclic-peptide that inhibits SARS-CoV-2 replication at
low nanomolar concentrations by the targeting of host protein eEF1A (eukaryotic translation-
elongation-factor-1A). We evaluated a model of intervention with plitidepsin in hospitalized
COVID-19 adult patients where three doses were assessed (1.5, 2 and 2.5 mg/day for 3 days, as a
90-minute intravenous infusion) in 45 patients (15 per dose-cohort). Treatment was well
5
tolerated, with only two Grade 3 treatment-related adverse events observed (hypersensitivity and
diarrhea). The discharge rates by Days 8 and 15 were 56.8% and 81.8%, respectively, with data
sustaining dose-effect. A mean 4.2 log10 viral load reduction was attained by Day 15.
Improvement in inflammation markers was also noted in a seemingly dose-dependent manner.
These results suggest that plitidepsin impacts the outcome of patients with COVID-19.
10
One-Sentence Summary: Plitidepsin, an inhibitor of SARS-Cov-2 in vitro, is safe and
positively influences the outcome of patients hospitalized with COVID-19.
15
Key Words: COVID-19 treatment, plitidepsin, eEF1A, SARS-CoV-2
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted May 25, 2021. ; https://doi.org/10.1101/2021.05.25.21257505doi: medRxiv preprint

4
Introduction
As of April 2021, there have been approximately 135 million confirmed cases of COVID-19
reported to the World Health Organization (WHO), including 3 million deaths. (1) The lack of
effective antiviral therapies represents a glaring unmet need, not only for the treatment of the
current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, (2) but also
5
for potential future pandemics, which may originate from other emergent coronaviruses. (3-5)
Viruses, particularly single-stranded positive-sense RNA viruses, are ubiquitous in the sea,
where they participate directly or indirectly in the population dynamics of marine organisms. (6)
Thus, the sea may prove to be a reservoir of potent, naturally selected antiviral compounds.
One such compound, plitidepsin, is a cyclic depsipeptide originally isolated from a
10
Mediterranean marine tunicate (Aplidium albicans) and is structurally related to didemnins, some
of which (i.e., those isolated from Trididemnum solidum) have shown antiviral properties. (7, 8)
Plitidepsin targets eukaryotic translation elongation factor 1A (eEF1A) (9), which is one of the
most abundant protein synthesis factors in the eukaryotic cell, (9, 10) and a protein known to be
used by viruses to facilitate their replication inside their host. (11) The SARS-CoV-2
15
nucleocapsid (N) protein is a key element involved in packaging of the viral RNA genome, (12,
13) and interacts with eEF1A. This interaction might be essential for viral replication, as eEF1A
knockdown results in a significant reduction in virus replication. (12, 13)
Originally developed as a cancer treatment, plitidepsin has undergone an extensive clinical
development program. Specifically, several phase I and II clinical trials have been conducted to
20
explore different intravenous dosing schedules and infusion times (14-18). Pharmacokinetic and
safety properties of plitidepsin were gathered from these studies. Based on the results obtained
from a phase III clinical trial (ADMYRE) (19), the Australian Therapeutic Goods Administration
(TGA) approved the combination of plitidepsin with dexamethasone for the treatment of patients
with relapsed/refractory multiple myeloma in 2018. (20)
25
In this study, we describe the results from a Proof-of Concept clinical trial that explores the
potential of plitidepsin as a therapy for patients hospitalized with COVID-19.
Results
We have evaluated the antiviral activity of plitidepsin against different coronavirus species,
30
strains and variants (Materials and Methods). Treatment of Huh-7 cells with as little as 0.5nM of
plitidepsin inhibited infection of a human coronavirus 229E expressing GFP (Figure 1A). A
10
4
- fold decrease in SARS-CoV genomic RNA (gRNA) accumulation and a 10
3
-fold decrease
in virus SARS-CoV titers were observed in VeroE6 cells treated with 50nM plitidepsin. (Figure
S1 and Table S1). By comparison, and consistent with previous results (10, 21) plitidepsin
35
showed nanomolar efficacy against SARS-CoV-2-induced cytopathic effects on Vero E6 cells
with a half-maximal inhibitory concentration (IC
50
) of 0.038µM, at concentrations where no
cytotoxic effects were observed (CC
50
2.9µM) (Figure 1B). Moreover, plitidpesin maintained its
nanomolar potency against replication of early as well as recently emerging SARS-CoV-2
lineages, such as B.1.1.7, in human lung and gastrointestinal cell lines (Figure 1C). Noteworthy,
40
plitidepsin was more effective against both variants than remdesivir (Figure 1C).
To identify target human plasma concentrations of plitidepsin for SARS-CoV-2 infection we
developed an extrapolation from in vitro results, in line with current recommendations. (22) This
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted May 25, 2021. ; https://doi.org/10.1101/2021.05.25.21257505doi: medRxiv preprint

5
approach integrated results from non-clinical studies described under Material and Methods
(Supplement), including in vitro drug sensitivity data for SARS-CoV-2 in Vero cells, human
plasma protein binding data (98%) (Table S2), and in vivo tissue distribution data in rats (lung-
to-plasma partition coefficient ratio of 543-fold (Table S3).
The target plasma and lung concentrations for plitidepsin were initially based on in vitro data
5
obtained by Boryung Pharmaceuticals, stablishing IC
50
of 3.26 nM and IC
90
of 9.38 nM
(Material & Methods). A validated pharmacokinetic population model of plitidepsin (23) was
used to simulate plasma exposures at different dose levels and infusion durations, so that
plitidepsin plasma profiles would reach 0.33 µg/L, and 0.96 µg/L, assuring target concentrations
in lung above the aforementioned in vitro IC
50
and IC
90
. A 3-day daily schedule was initially
10
selected to achieve sustained active exposures, under the hypothesis that an acute reduction of
the viral load would prevent the onset of the more severe inflammatory phase of COVID-19. The
predicted plasma concentrations of plitidepsin, at a dose of 1.5 mg infused intravenously over
90 min, were above the target IC
50
for the full treatment period and above the IC
90
for half of the
treatment period. The respective predictions after a dose of 2.5 mg were above the IC
90
during
15
most of the treatment period (Figure 2). Thus, we anticipated that the proposed range of doses
would result in stable active concentrations in critical anatomical compartments, such as lung,
for more than 120 hours. This model was later supported by White et al, who reported an IC
90
of
0.88 nM (10). To reach this target concentration in lung tissue, according to the above reasoning,
plitidepsin plasma concentration should be above 0.18 µg/L.
20
We subsequently designed the APLICOV-PC study [APL-D-002-20; EudraCT #2020-001993-
31; NCT #04382066] as a proof-of-concept clinical trial, exploring three dose levels of
plitidepsin (1.5 mg/day, 2.0 mg/day, and 2.5 mg/day, flat doses) for 3 consecutive days, as a 90-
min intravenous (IV) infusion, in adult patients with COVID-19 who required hospitalization.
The primary endpoints were related to safety, but secondary efficacy and pharmacodynamic
25
endpoints were also included (Material and Methods).
Enrollment began on May 12, 2020 and the study was completed by November 26, 2020. Here,
we present the data analyses by the cut-off date of December 10, 2020.
In total, 46 hospitalized COVID-19 patients were enrolled across 10 sites in Spain. Baseline
demographic and clinical characteristics are summarized in Table 1. One patient withdrew
30
consent before initiating study-specific procedures. The 45 patients who received treatment were
sequentially allocated to one of the three dose cohorts; whenever more than 1 dose cohort was
open, a patient was assigned by central randomization (Material and Methods).
Forty-four patients completed the study through day 31, with one patient in the 1.5 mg cohort
withdrawing from the study after the first infusion due to a grade 3 hypersensitivity reaction. The
35
average patient age was 52 years (range 3184 years). Most patients were male (66.7%) and 80%
had co-morbidities (46.7% had two or more). The most commonly reported comorbidities were
obesity (22.2%), hypertension (20%), and type 2 diabetes mellitus (17.8%). The distribution of
comorbidities was similar between the three treatment cohorts.
Most patients had moderate COVID-19 (51.1%), according to FDA categorization (24), with
40
13.3% and 35.6% having mild and severe disease, respectively. Baseline chest X-rays showed
evidence of lower respiratory infection (infiltrates, unilateral pneumonia, or bilateral pneumonia)
in 41 of 45 treated patients (91%), with bilateral pneumonia seen in 32 of them (71%); the
percentage of patients with bilateral pneumonia was similar across dose cohorts. Viral load was
similar across the three cohorts, with average baseline values for SARS-CoV-2 RNA from
45
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted May 25, 2021. ; https://doi.org/10.1101/2021.05.25.21257505doi: medRxiv preprint

Citations
More filters
Journal ArticleDOI
TL;DR: Control of the COVID-19 pandemic will require the use of pharmacological treatments against SARS-CoV2, and drugs that address the hyperinflammatory phase of the disease triggered by the so-called cytokine storm are particularly relevant.
Abstract: In spite of advances in vaccination, control of the COVID-19 pandemic will require the use of pharmacological treatments against SARS-CoV2. Their development needs to consider the existence of two phases in the disease, namely the viral infection and the inflammatory stages. The main targets for antiviral therapeutic intervention are: (a) viral proteins, including the spike (S) protein characteristic of the viral cover and the viral proteases in charge of processing the polyprotein arising from viral genome translation; (b) host proteins, such as those involved in the processes related to viral entry into the host cell and the release of the viral genome inside the cell, the elongation factor eEF1A and importins. The use of antivirals targeted at host proteins is less developed but it has the potential advantage of not being affected by mutations in the genome of the virus and therefore being active against all its variants. Regarding drugs that address the hyperinflammatory phase of the disease triggered by the so-called cytokine storm, the following strategies are particularly relevant: (a) drugs targeting JAK kinases; (b) sphingosine kinase 2 inhibitors; (c) antibodies against interleukin 6 or its receptor; (d) use of the traditional anti-inflammatory corticosteroids.

19 citations

Journal ArticleDOI
TL;DR: A 75-year-old male with chronic lymphocytic leukemia who was deficient in CD19+CD20+ B-lymphocyte populations due to previous treatment with anti-CD20 monoclonal antibodies was treated with two courses of the antiviral plitidepsin on a compassionate use basis as mentioned in this paper .
Abstract: There is an urgent need for highly efficacious antiviral therapies in immunosuppressed hosts who develop coronavirus disease (COVID-19), with special concern for those affected by hematological malignancies.Here, we report the case of a 75-year-old male with chronic lymphocytic leukemia who was deficient in CD19+CD20+ B-lymphocyte populations due to previous treatment with anti-CD20 monoclonal antibodies. The patient presented with severe COVID-19 pneumonia due to prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and was treated with two courses of the antiviral plitidepsin on a compassionate use basis. The patient subsequently achieved an undetectable viral load, and his pneumonia resolved.Treatment with plitidepsin was well-tolerated without any further hematological or cardiovascular toxicities. This case further supports plitidepsin as a potential antiviral drug in SARS-CoV-2 patients affected by immune deficiencies and hematological malignancies.

12 citations

Journal ArticleDOI
TL;DR: In this paper, the authors argue that although COVID-19 was initially considered a new challenge, justifying extraordinary response measures, this situation has changed -and so should our response.

3 citations

Journal ArticleDOI
TL;DR: In this paper, a patient with stage IIIB gastric signet ring cell carcinoma and multiple comorbidities developed Coronavirus disease (COVID-19) shortly after receiving his first chemotherapy course.

1 citations

Journal ArticleDOI
TL;DR: In this article , the authors assessed the recent literature on drugs under advanced clinical evaluation for COVID-19 with broad spectrum activity, including antiviral agents and host-directed therapies (HDTs), which may be relevant for future coping with Biological Warfare Agents (BWAs), as well as with other agents, in particular respiratory infections.
Abstract: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak resulted in hundreds of millions of coronavirus cases, as well as millions of deaths worldwide. Coronavirus Disease 2019 (COVID-19), the disease resulting from exposure to this pathogen, is characterized, among other features, by a pulmonary pathology, which can progress to “cytokine storm”, acute respiratory distress syndrome (ARDS), respiratory failure and death. Vaccines are the unsurpassed strategy for prevention and protection against the SARS-CoV-2 infection. However, there is still an extremely high number of severely ill people from at-risk populations. This may be attributed to waning immune response, variant-induced breakthrough infections, unvaccinated population, etc. It is therefore of high importance to utilize pharmacological-based treatments, despite the progression of the global vaccination campaign. Until the approval of Paxlovid, an efficient and highly selective anti-SARS-CoV-2 drug, and the broad-spectrum antiviral agent Lagevrio, many pharmacological-based countermeasures were, and still are, being evaluated in clinical trials. Some of these are host-directed therapies (HDTs), which modulate the endogenic response against the virus, and therefore may confer efficient protection against a wide array of pathogens. These could potentially include Biological Warfare Agents (BWAs), exposure to which may lead to mass casualties due to disease severity and a possible lack of efficient treatment. In this review, we assessed the recent literature on drugs under advanced clinical evaluation for COVID-19 with broad spectrum activity, including antiviral agents and HDTs, which may be relevant for future coping with BWAs, as well as with other agents, in particular respiratory infections.
References
More filters
Journal ArticleDOI
David E. Gordon, Gwendolyn M. Jang, Mehdi Bouhaddou, Jiewei Xu, Kirsten Obernier, Kris M. White1, Matthew J. O’Meara2, Veronica V. Rezelj3, Jeffrey Z. Guo, Danielle L. Swaney, Tia A. Tummino4, Ruth Hüttenhain, Robyn M. Kaake, Alicia L. Richards, Beril Tutuncuoglu, Helene Foussard, Jyoti Batra, Kelsey M. Haas, Maya Modak, Minkyu Kim, Paige Haas, Benjamin J. Polacco, Hannes Braberg, Jacqueline M. Fabius, Manon Eckhardt, Margaret Soucheray, Melanie J. Bennett, Merve Cakir, Michael McGregor, Qiongyu Li, Bjoern Meyer3, Ferdinand Roesch3, Thomas Vallet3, Alice Mac Kain3, Lisa Miorin1, Elena Moreno1, Zun Zar Chi Naing, Yuan Zhou, Shiming Peng4, Ying Shi, Ziyang Zhang, Wenqi Shen, Ilsa T Kirby, James E. Melnyk, John S. Chorba, Kevin Lou, Shizhong Dai, Inigo Barrio-Hernandez5, Danish Memon5, Claudia Hernandez-Armenta5, Jiankun Lyu4, Christopher J.P. Mathy, Tina Perica4, Kala Bharath Pilla4, Sai J. Ganesan4, Daniel J. Saltzberg4, Rakesh Ramachandran4, Xi Liu4, Sara Brin Rosenthal6, Lorenzo Calviello4, Srivats Venkataramanan4, Jose Liboy-Lugo4, Yizhu Lin4, Xi Ping Huang7, Yongfeng Liu7, Stephanie A. Wankowicz, Markus Bohn4, Maliheh Safari4, Fatima S. Ugur, Cassandra Koh3, Nastaran Sadat Savar3, Quang Dinh Tran3, Djoshkun Shengjuler3, Sabrina J. Fletcher3, Michael C. O’Neal, Yiming Cai, Jason C.J. Chang, David J. Broadhurst, Saker Klippsten, Phillip P. Sharp4, Nicole A. Wenzell4, Duygu Kuzuoğlu-Öztürk4, Hao-Yuan Wang4, Raphael Trenker4, Janet M. Young8, Devin A. Cavero9, Devin A. Cavero4, Joseph Hiatt9, Joseph Hiatt4, Theodore L. Roth, Ujjwal Rathore9, Ujjwal Rathore4, Advait Subramanian4, Julia Noack4, Mathieu Hubert3, Robert M. Stroud4, Alan D. Frankel4, Oren S. Rosenberg, Kliment A. Verba4, David A. Agard4, Melanie Ott, Michael Emerman8, Natalia Jura, Mark von Zastrow, Eric Verdin10, Eric Verdin4, Alan Ashworth4, Olivier Schwartz3, Christophe d'Enfert3, Shaeri Mukherjee4, Matthew P. Jacobson4, Harmit S. Malik8, Danica Galonić Fujimori, Trey Ideker6, Charles S. Craik, Stephen N. Floor4, James S. Fraser4, John D. Gross4, Andrej Sali, Bryan L. Roth7, Davide Ruggero, Jack Taunton4, Tanja Kortemme, Pedro Beltrao5, Marco Vignuzzi3, Adolfo García-Sastre, Kevan M. Shokat, Brian K. Shoichet4, Nevan J. Krogan 
30 Apr 2020-Nature
TL;DR: A human–SARS-CoV-2 protein interaction map highlights cellular processes that are hijacked by the virus and that can be targeted by existing drugs, including inhibitors of mRNA translation and predicted regulators of the sigma receptors.
Abstract: A newly described coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of coronavirus disease 2019 (COVID-19), has infected over 2.3 million people, led to the death of more than 160,000 individuals and caused worldwide social and economic disruption1,2. There are no antiviral drugs with proven clinical efficacy for the treatment of COVID-19, nor are there any vaccines that prevent infection with SARS-CoV-2, and efforts to develop drugs and vaccines are hampered by the limited knowledge of the molecular details of how SARS-CoV-2 infects cells. Here we cloned, tagged and expressed 26 of the 29 SARS-CoV-2 proteins in human cells and identified the human proteins that physically associated with each of the SARS-CoV-2 proteins using affinity-purification mass spectrometry, identifying 332 high-confidence protein–protein interactions between SARS-CoV-2 and human proteins. Among these, we identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration, 12 are in clinical trials and 28 are preclinical compounds). We screened a subset of these in multiple viral assays and found two sets of pharmacological agents that displayed antiviral activity: inhibitors of mRNA translation and predicted regulators of the sigma-1 and sigma-2 receptors. Further studies of these host-factor-targeting agents, including their combination with drugs that directly target viral enzymes, could lead to a therapeutic regimen to treat COVID-19. A human–SARS-CoV-2 protein interaction map highlights cellular processes that are hijacked by the virus and that can be targeted by existing drugs, including inhibitors of mRNA translation and predicted regulators of the sigma receptors.

3,319 citations

Journal ArticleDOI
TL;DR: A potential risk of SARS-CoV re-emergence from viruses currently circulating in bat populations is suggested, and robust viral replication both in vitro and in vivo is demonstrated.
Abstract: The emergence of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome (MERS)-CoV underscores the threat of cross-species transmission events leading to outbreaks in humans. Here we examine the disease potential of a SARS-like virus, SHC014-CoV, which is currently circulating in Chinese horseshoe bat populations. Using the SARS-CoV reverse genetics system, we generated and characterized a chimeric virus expressing the spike of bat coronavirus SHC014 in a mouse-adapted SARS-CoV backbone. The results indicate that group 2b viruses encoding the SHC014 spike in a wild-type backbone can efficiently use multiple orthologs of the SARS receptor human angiotensin converting enzyme II (ACE2), replicate efficiently in primary human airway cells and achieve in vitro titers equivalent to epidemic strains of SARS-CoV. Additionally, in vivo experiments demonstrate replication of the chimeric virus in mouse lung with notable pathogenesis. Evaluation of available SARS-based immune-therapeutic and prophylactic modalities revealed poor efficacy; both monoclonal antibody and vaccine approaches failed to neutralize and protect from infection with CoVs using the novel spike protein. On the basis of these findings, we synthetically re-derived an infectious full-length SHC014 recombinant virus and demonstrate robust viral replication both in vitro and in vivo. Our work suggests a potential risk of SARS-CoV re-emergence from viruses currently circulating in bat populations.

762 citations

Journal ArticleDOI
TL;DR: It is reported that a higher prevalence of detectable SARS-CoV-2 plasma viral load is associated with worse respiratory disease severity, lower absolute lymphocyte counts, and increased markers of inflammation, including C-reactive protein and IL-6.
Abstract: The relationship between SARS-CoV-2 viral load and risk of disease progression remains largely undefined in coronavirus disease 2019 (COVID-19). Here, we quantify SARS-CoV-2 viral load from participants with a diverse range of COVID-19 disease severity, including those requiring hospitalization, outpatients with mild disease, and individuals with resolved infection. We detected SARS-CoV-2 plasma RNA in 27% of hospitalized participants, and 13% of outpatients diagnosed with COVID-19. Amongst the participants hospitalized with COVID-19, we report that a higher prevalence of detectable SARS-CoV-2 plasma viral load is associated with worse respiratory disease severity, lower absolute lymphocyte counts, and increased markers of inflammation, including C-reactive protein and IL-6. SARS-CoV-2 viral loads, especially plasma viremia, are associated with increased risk of mortality. Our data show that SARS-CoV-2 viral loads may aid in the risk stratification of patients with COVID-19, and therefore its role in disease pathogenesis should be further explored.

595 citations

Journal ArticleDOI
TL;DR: An approach that combines existing metagenomics data with reverse genetics to engineer reagents to evaluate emergence and pathogenic potential of circulating zoonotic viruses indicates that the WIV1-coronavirus (CoV) cluster has the ability to directly infect and may undergo limited transmission in human populations.
Abstract: Outbreaks from zoonotic sources represent a threat to both human disease as well as the global economy. Despite a wealth of metagenomics studies, methods to leverage these datasets to identify future threats are underdeveloped. In this study, we describe an approach that combines existing metagenomics data with reverse genetics to engineer reagents to evaluate emergence and pathogenic potential of circulating zoonotic viruses. Focusing on the severe acute respiratory syndrome (SARS)-like viruses, the results indicate that the WIV1-coronavirus (CoV) cluster has the ability to directly infect and may undergo limited transmission in human populations. However, in vivo attenuation suggests additional adaptation is required for epidemic disease. Importantly, available SARS monoclonal antibodies offered success in limiting viral infection absent from available vaccine approaches. Together, the data highlight the utility of a platform to identify and prioritize prepandemic strains harbored in animal reservoirs and document the threat posed by WIV1-CoV for emergence in human populations.

361 citations

Frequently Asked Questions (8)
Q1. What are the contributions in "Plitidepsin has a positive therapeutic index in adult patients with covid-19 requiring hospitalization" ?

Authors: José F. Varona, Pedro Landete, Jose A. Lopez-Martin, Vicente Estrada, Roger Paredes, Pablo Guisado-Vasco, Lucía Fernández de Orueta, Miguel Torralba, Jesús 5 Fortún, Roberto Vates, José Barberán, Bonaventura Clotet, Julio Ancochea, Daniel Carnevali, Noemí Cabello, Lourdes Porras, Paloma Gijón, Alfonso Monereo, Daniel Abad, Sonia Zúñiga, Isabel Sola, Jordi Rodon, Nuria Izquierdo-Useros, Salvador Fudio, María José Pontes, Beatriz de Rivas, Patricia Girón de Velasco, Belén Sopesén, Antonio Nieto, Javier Gómez, Pablo Avilés, Rubin Lubomirov, Kris M. 10 White, Romel Rosales, Soner Yildiz, Ann-Kathrin Reuschl, Lucy G. Thorne, Clare Jolly, Greg J. Towers, Lorena Zuliani-Alvarez, Mehdi Bouhaddou, Kirsten Obernier, Luis Enjuanes, Jose M. Fernández-Sousa, Plitidepsin – COVID 19 Study Group, Nevan J. Krogan, José M. Jimeno, Adolfo García-Sastre. 

(11) The SARS-CoV-2 15nucleocapsid (N) protein is a key element involved in packaging of the viral RNA genome, (12, 13) and interacts with eEF1A. 

The authors hypothesize that plitidepsin, besides acting as an antiviral agent, may also modulate immune response by its effects on monocytes/macrophages. 

particularly single-stranded positive-sense RNA viruses, are ubiquitous in the sea, where they participate directly or indirectly in the population dynamics of marine organisms. 

As of April 2021, there have been approximately 135 million confirmed cases of COVID-19 reported to the World Health Organization (WHO), including 3 million deaths. 

plitidpesin maintained its nanomolar potency against replication of early as well as recently emerging SARS-CoV-2lineages, such as B.1.1.7, in human lung and gastrointestinal cell lines (Figure 1C). 

Given the small sample size, the baseline variables the authors selected for exploratory purposes were limited to age, viral load, diseaseseverity, and dose cohort. 

In summary, the authors have integrated preclinical and clinical studies onthe use of plitidepsin to treat SARS-CoV-2 and other coronavirus infections, and generated 25promising patient data supporting the launching of phase 3 clinical studies to demonstrate the efficacy of treatment with plitidepsin in moderate COVID-19 patients.